Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 220(8)2023 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-37158982

RESUMO

Type I interferons are important antiviral cytokines, but prolonged interferon production is detrimental to the host. The TLR3-driven immune response is crucial for mammalian antiviral immunity, and its intracellular localization determines induction of type I interferons; however, the mechanism terminating TLR3 signaling remains obscure. Here, we show that the E3 ubiquitin ligase ZNRF1 controls TLR3 sorting into multivesicular bodies/lysosomes to terminate signaling and type I interferon production. Mechanistically, c-Src kinase activated by TLR3 engagement phosphorylates ZNRF1 at tyrosine 103, which mediates K63-linked ubiquitination of TLR3 at lysine 813 and promotes TLR3 lysosomal trafficking and degradation. ZNRF1-deficient mice and cells are resistant to infection by encephalomyocarditis virus and SARS-CoV-2 because of enhanced type I interferon production. However, Znrf1-/- mice have exacerbated lung barrier damage triggered by antiviral immunity, leading to enhanced susceptibility to respiratory bacterial superinfections. Our study highlights the c-Src-ZNRF1 axis as a negative feedback mechanism controlling TLR3 trafficking and the termination of TLR3 signaling.


Assuntos
COVID-19 , Interferon Tipo I , Animais , Camundongos , Antivirais , SARS-CoV-2 , Receptor 3 Toll-Like , Genes src
2.
Inorg Chem ; 61(9): 3997-4008, 2022 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-35020371

RESUMO

In this study, aluminum complexes bearing ferrocene-based and arylthiomethylphenolate ligands were synthesized, and their catalytic activity for ε-caprolactone (CL) polymerization was investigated. The catalytic activity of the reduced form of Al complexes was higher than that of the oxidized form. The CL polymerization rate of the reduced form fcO2AlMe (75 min, conversion = 100%) was higher than that of the oxidized form fcoxO2AlMe (4320 min, conversion = 45%), and the CL polymerization rate of fc(OAlMe2)2 (40 min, conversion = 100%) was higher than that of fcox(OAlMe2)2 (60 min, conversion = 97%). Electron deficiency substituents on phenolate decreased the catalytic activity of Al complexes bearing arylthiomethylphenolate ligands. Density functional theory calculations revealed that thioether coordination stabilized the transition state (TS1) and that the oxidized form fcox(OAlMe2)2 exhibited weaker thioether coordination and higher activation energy in TS1 compared with those of the reduced form fcO2AlMe. In addition, our study determined that the thioether group is a suitable chelating group for Al catalysts in CL polymerization due to its labile nature.

3.
Adv Exp Med Biol ; 1204: 197-214, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32152948

RESUMO

Siglecs are sialic acid (Sia) recognizing immunoglobulin-like receptors expressed on the surface of all the major leukocyte lineages in mammals. Siglecs recognize ubiquitous Sia epitopes on various glycoconjugates in the cell glycocalyx and transduce signals to regulate immunological and inflammatory activities of these cells. The subset known as CD33-related Siglecs is principally inhibitory receptors that suppress leukocyte activation, and recent research has shown that a number of bacterial pathogens use Sia mimicry to engage these Siglecs as an immune evasion strategy. Conversely, Siglec-1 is a macrophage phagocytic receptor that engages GBS and other sialylated bacteria to promote effective phagocytosis and antigen presentation for the adaptive immune response, whereas certain viruses and parasites use Siglec-1 to gain entry to immune cells as a proximal step in the infectious process. Siglecs are positioned in crosstalk with other host innate immune sensing pathways to modulate the immune response to infection in complex ways. This chapter summarizes the current understanding of Siglecs at the host-pathogen interface, a field of study expanding in breadth and medical importance, and which provides potential targets for immune-based anti-infective strategies.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/imunologia , Animais , Glicocálix/imunologia , Humanos , Leucócitos/citologia , Leucócitos/imunologia , Macrófagos/imunologia , Fagocitose , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-29616195

RESUMO

Clostridium difficile infection (CDI) is the leading cause of nosocomial infection in hospitalized patients receiving long-term antibiotic treatment. An excessive host inflammatory response is believed to be the major mechanism underlying the pathogenesis of C. difficile infection, and various proinflammatory cytokines such as IL-1ß are detected in patients with C. difficile infection. IL-1ß is known to be processed by caspase-1, a cysteine protease that is regulated by a protein complex called the inflammasome, which leads to a specialized form of cell death called pyroptosis. The function of inflammasome activation-induced pyroptosis is to clear or limit the spread of invading pathogens via infiltrated neutrophils. Here, we focused on inflammasome activation induced by intact C. difficile to re-evaluate the nature of inflammasome activation in CDI pathogenesis, which could provide information that leads to an alternative therapeutic strategy for the treatment of this condition in humans. First, we found that caspase-1-dependent IL-1ß production was induced by C. difficile pathogens in macrophages and increased in a time-dependent manner. Moreover, intracellular toxigenic C. difficile was essential for ATP-P2X7 pathway of inflammasome activation and subsequent caspase-1-dependent pyroptotic cell death, leading to the loss of membrane integrity and release of intracellular contents such as LDH. Notably, we also observed that bacterial components such as surface layer proteins (SLPs) were released from pyroptotic cells. In addition, pro-IL-1ß production was completely MyD88 and partially TLR2 dependent. Finally, to investigate the role of the caspase-1-dependent inflammasome in host defense, we found that colonic inflammasome activation was also induced by CDI and that caspase-1 inhibition by Ac-YVAD-CMK led to increased disease progression and C. difficile load. Taken together, the present results suggest that MyD88 and TLR2 are critical component in pro-IL-1ß production and intracellular C. difficile following the ATP-P2X7 pathway of inflammasome activation and pyroptosis, which play important roles in host defense through the utilization of inflammation-mediated bacterial clearance mechanisms during C. difficile infection.


Assuntos
Trifosfato de Adenosina/imunologia , Clostridioides difficile/imunologia , Infecções por Clostridium/imunologia , Inflamassomos/imunologia , Receptores Purinérgicos P2X7/imunologia , Animais , Caspase 1/genética , Caspase 1/imunologia , Clostridioides difficile/genética , Infecções por Clostridium/genética , Infecções por Clostridium/microbiologia , Humanos , Inflamassomos/genética , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Purinérgicos P2X7/genética , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia
5.
Nat Commun ; 8: 15502, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28593998

RESUMO

Caveolin-1 (CAV1), the major constituent of caveolae, plays a pivotal role in various cellular biological functions, including cancer and inflammation. The ubiquitin/proteasomal pathway is known to contribute to the regulation of CAV1 expression, but the ubiquitin ligase responsible for CAV1 protein stability remains unidentified. Here we reveal that E3 ubiquitin ligase ZNRF1 modulates CAV1 protein stability to regulate Toll-like receptor (TLR) 4-triggered immune responses. We demonstrate that ZNRF1 physically interacts with CAV1 in response to lipopolysaccharide and mediates ubiquitination and degradation of CAV1. The ZNRF1-CAV1 axis regulates Akt-GSK3ß activity upon TLR4 activation, resulting in enhanced production of pro-inflammatory cytokines and inhibition of anti-inflammatory cytokine IL-10. Mice with deletion of ZNRF1 in their hematopoietic cells display increased resistance to endotoxic and polymicrobial septic shock due to attenuated inflammation. Our study defines ZNRF1 as a regulator of TLR4-induced inflammatory responses and reveals another mechanism for the regulation of TLR4 signalling through CAV1.


Assuntos
Caveolina 1/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Sequência de Aminoácidos , Animais , Caveolina 1/química , Ceco/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Deleção de Genes , Glicogênio Sintase Quinase 3 beta/metabolismo , Mediadores da Inflamação/metabolismo , Ligadura , Lipopolissacarídeos , Lisina/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Punções , Células RAW 264.7 , Choque Séptico/imunologia , Choque Séptico/metabolismo , Choque Séptico/patologia , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Ubiquitina-Proteína Ligases/deficiência
6.
Glycobiology ; 24(9): 818-25, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24996821

RESUMO

Siglecs are mammalian sialic acid (Sia) recognizing immunoglobulin-like receptors expressed across the major leukocyte lineages, and function to recognize ubiquitous Sia epitopes on cell surface glycoconjugates and regulate immunological and inflammatory activities of these cells. A large subset referred to as CD33-related Siglecs are inhibitory receptors that limit leukocyte activation, and recent research has shown that the pathogen group B Streptococcus (GBS) binds to these Siglecs in Sia- and protein-dependent fashion to downregulate leukocyte bactericidal capacity. Conversely, sialoadhesin is a macrophage phagocytic receptor that engages GBS and other sialylated pathogens to promote effective phagocytosis and antigen presentation for the adaptive immune response. A variety of other important Siglec interactions with bacterial, viral and protozoan pathogens are beginning to be recognized. Siglec genes and binding specificities are rapidly evolving among primates, with key extant polymorphisms in human populations that may influence susceptibility to infection-associated disorders including chronic obstructive pulmonary disease and premature birth. This review summarizes current understanding of interactions between pathogens and Siglecs, a field of investigation that is likely to continue expanding in scope and medical importance.


Assuntos
HIV/imunologia , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/imunologia , Ácidos Siálicos/metabolismo , Streptococcus/imunologia , Animais , Evolução Molecular , Humanos , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/metabolismo
7.
J Mol Med (Berl) ; 92(9): 951-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24788876

RESUMO

UNLABELLED: Several bacterial pathogens decorate their surfaces with sialic acid (Sia) residues within cell wall components or capsular exopolysaccharides. Sialic acid expression can promote bacterial virulence by blocking complement activation or by engagement of inhibitory sialic acid-binding immunoglobulin-like lectins (Siglecs) on host leukocytes. Expressed at high levels on splenic and lymph node macrophages, sialoadhesin (Sn) is a unique Siglec with an elongated structure that lacks intracellular signaling motifs. Sialoadhesin allows macrophage to engage certain sialylated pathogens and stimulate inflammatory responses, but the in vivo significance of sialoadhesin in infection has not been shown. We demonstrate that macrophages phagocytose the sialylated pathogen group B Streptococcus (GBS) and increase bactericidal activity via sialoadhesin-sialic-acid-mediated recognition. Sialoadhesin expression on marginal zone metallophilic macrophages in the spleen trapped circulating GBS and restricted the spread of the GBS to distant organs, reducing mortality. Specific IgM antibody responses to GBS challenge were also impaired in sialoadhesin-deficient mice. Thus, sialoadhesin represents a key bridge to orchestrate innate and adaptive immune defenses against invasive sialylated bacterial pathogens. KEY MESSAGE: Sialoadhesin is critical for macrophages to phagocytose and clear GBS. Increased GBS organ dissemination in the sialoadhesin-deficient mice. Reduced anti-GBS IgM production in the sialoadhesin-deficient mice.


Assuntos
Interações Hospedeiro-Patógeno , Macrófagos/microbiologia , Fagocitose , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus agalactiae/fisiologia , Animais , Células Cultivadas , Deleção de Genes , Imunoglobulina M/imunologia , Macrófagos/imunologia , Camundongos , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Streptococcus agalactiae/imunologia
8.
PLoS Pathog ; 10(1): e1003846, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24391502

RESUMO

Group B Streptococcus (GBS) is a common agent of bacterial sepsis and meningitis in newborns. The GBS surface capsule contains sialic acids (Sia) that engage Sia-binding immunoglobulin-like lectins (Siglecs) on leukocytes. Here we use mice lacking Siglec-E, an inhibitory Siglec of myelomonocytic cells, to study the significance of GBS Siglec engagement during in vivo infection. We found GBS bound to Siglec-E in a Sia-specific fashion to blunt NF-κB and MAPK activation. As a consequence, Siglec-E-deficient macrophages had enhanced pro-inflammatory cytokine secretion, phagocytosis and bactericidal activity against the pathogen. Following pulmonary or low-dose intravenous GBS challenge, Siglec-E KO mice produced more pro-inflammatory cytokines and exhibited reduced GBS invasion of the central nervous system. In contrast, upon high dose lethal challenges, cytokine storm in Siglec-E KO mice was associated with accelerated mortality. We conclude that GBS Sia mimicry influences host innate immune and inflammatory responses in vivo through engagement of an inhibitory Siglec, with the ultimate outcome of the host response varying depending upon the site, stage and magnitude of infection.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos B/imunologia , Mimetismo Molecular/imunologia , Ácido N-Acetilneuramínico/imunologia , Pneumonia Bacteriana/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus agalactiae/imunologia , Animais , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos B/genética , Citocinas/genética , Citocinas/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Ácido N-Acetilneuramínico/genética , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/patologia , Infecções Estreptocócicas/genética , Infecções Estreptocócicas/patologia
9.
J Immunol ; 188(12): 5867-76, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22581854

RESUMO

Decoy receptor 3 (DcR3) is a soluble protein in the TNFR superfamily. Its known ligands include Fas ligand, homologous to lymphotoxin, showing inducible expression, and competing with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes, TNF-like molecule 1A, and heparan sulfate proteoglycans. DcR3 has been reported to modulate the functions of T cells, dendritic cells, and macrophages; however, its role in regulating B cell activation is largely unknown. In this study, we found that the DcR3.Fc fusion protein bound to human and mouse B cells and suppressed the activation of B cells. DcR3.Fc attenuated Staphylococcus aureus, IgM-, Pam(3)CSK(4)-, and LPS-mediated B cell proliferation but did not affect cytokine-induced B cell growth. In the presence of these mitogens, DcR3.Fc did not induce B cell apoptosis, suggesting that DcR3 may inhibit the signal(s) important for B cell activation. Because the combination of Fas.Fc, LT-ßR.Fc (homologous to lymphotoxin, showing inducible expression, and competing with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes receptor), and DR3.Fc (TNF-like molecule 1A receptor) did not suppress B cell proliferation and because the biological effect of DcR3.Fc on B cells was not blocked by heparin, we hypothesize that a novel ligand(s) of DcR3 mediates its inhibitory activity on B cells. Moreover, we found that TLR2-stimulated NF-κB p65 activation and NF-κB-driven luciferase activity were attenuated by DcR3.Fc. The TLR2-induced cytokine production by B cells was consistently reduced by DcR3. These results imply that DcR3 may regulate B cell activation by suppressing the activation of NF-κB.


Assuntos
Linfócitos B/imunologia , Ativação Linfocitária/imunologia , NF-kappa B/imunologia , Membro 6b de Receptores do Fator de Necrose Tumoral/imunologia , Receptor 2 Toll-Like/imunologia , Animais , Apoptose/imunologia , Linfócitos B/metabolismo , Proliferação de Células , Células Cultivadas , Humanos , Camundongos , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Membro 6b de Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes de Fusão , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/metabolismo
10.
Cancer Sci ; 102(4): 895-902, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21219537

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is one prevalent human cancer worldwide. No molecular markers are presently used for predicting prognosis in HNSCC. Krüppel-like factor 4 (KLF4) is a transcription factor with diverse physiological functions, and possesses opposing roles in different human cancers. The expression and roles of KLF4 in HNSCC remain to be elucidated. In this study, immunohistochemical (IHC) analysis of KLF4 in 62 HNSCC was firstly performed. IHC results demonstrated that 42 (67.7%) had decreased KLF4 expression compared with surrounding normal epithelium, while persistent KLF4 expression was demonstrated in 20 (32.3%). The IHC results were further verified by Western blot and real-time PCR analyses to confirm the robustness of staining and interpretation. Interestingly, persistent KLF4 expression independently correlated with a worse disease-specific survival (P = 0.005), especially in patients with advanced disease. In consistent with clinical observation, all five HNSCC cell lines tested revealed a low level of baseline KLF4 expression. Moreover, enforced KLF4 expression in cell line SAS significantly increased in vitro migration/invasion abilities, multi-drug resistance, and in vivo tumorigenicity. These results clearly illustrate that persistent KLF4 expression predicts poor prognosis and confers aggressiveness in HNSCC. Our data therefore provides valuable information that HNSCC with persistent KLF4 expression might require intensified combination treatment in future practice.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Recidiva Local de Neoplasia/metabolismo , Animais , Western Blotting , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/secundário , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisplatino/administração & dosagem , Progressão da Doença , Docetaxel , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Fluoruracila/administração & dosagem , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Técnicas Imunoenzimáticas , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Taxoides/administração & dosagem , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nat Biotechnol ; 27(6): 567-71, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19448630

RESUMO

RNA interference (RNAi) induced by short interfering RNA (siRNA) allows for discovery research and large-scale screening; however, owing to their size and anionic charge, siRNAs do not readily enter cells. Current approaches do not deliver siRNAs into a high percentage of primary cells without cytotoxicity. Here we report an efficient siRNA delivery approach that uses a peptide transduction domain-double-stranded RNA-binding domain (PTD-DRBD) fusion protein. DRBDs bind to siRNAs with high avidity, masking the siRNA's negative charge and allowing PTD-mediated cellular uptake. PTD-DRBD-delivered siRNA induced rapid RNAi in a large percentage of various primary and transformed cells, including T cells, human umbilical vein endothelial cells and human embryonic stem cells. We observed no cytotoxicity, minimal off-target transcriptional changes and no induction of innate immune responses. Thus, PTD-DRBD-mediated siRNA delivery allows efficient gene silencing in difficult-to-transfect primary cell types.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Ligação a RNA/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células-Tronco Embrionárias , Células Epiteliais , Citometria de Fluxo , Gliceraldeído-3-Fosfato Desidrogenases/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células Jurkat , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas de Membrana Transportadoras/genética , Camundongos , Estrutura Terciária de Proteína , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T , Veias Umbilicais , Proteína Vermelha Fluorescente
12.
Blood ; 111(10): 5054-63, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18349319

RESUMO

Decoy receptor 3 (DcR3) is a member of the TNF receptor superfamily and is up-regulated in tumors originating from a diversity of lineages. DcR3 is capable of promoting angiogenesis, inducing dendritic cell apoptosis, and modulating macrophage differentiation. Since tumor-associated macrophages (TAMs) are the major infiltrating leukocytes in most malignant tumors, we used microarray technology to investigate whether DcR3 contributes to the development of TAMs. Among the DcR3-modulated genes expressed by TAMs, those that encode proteins involved in MHC class II (MHC-II)-dependent antigen presentation were down-regulated substantially, together with the master regulator of MHC-II expression (the class II transactivator, CIITA). The ERK- and JNK-induced deacetylation of histones associated with the CIITA promoters was responsible for DcR3-mediated down-regulation of MHC-II expression. Furthermore, the expression level of DcR3 in cancer cells correlated inversely with HLA-DR levels on TAMs and with the overall survival time of pancreatic cancer patients. The role of DcR3 in the development of TAMs was further confirmed using transgenic mice overexpressing DcR3. This elucidates the molecular mechanism of impaired MHC-II-mediated antigen presentation by TAMs, and raises the possibility that subversion of TAM-induced immunosuppression via inhibition of DcR3 expression might represent a target for the design of new therapeutics.


Assuntos
Epigênese Genética , Genes MHC da Classe II/genética , Macrófagos/metabolismo , Neoplasias/imunologia , Proteínas Nucleares/genética , Membro 6b de Receptores do Fator de Necrose Tumoral/fisiologia , Transativadores/genética , Animais , Apresentação de Antígeno/genética , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas
13.
Blood ; 111(3): 1480-8, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18006694

RESUMO

Decoy receptor 3 (DcR3) is a soluble decoy receptor belonging to the tumor necrosis factor receptor (TNFR) superfamily, and its expression is not only up-regulated in cancer cells derived from various cell lineages, but also correlates with overall survival of patients with cancer. It has been shown that DcR3 sensitize cells of hematopoietic origin to TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis; therefore, we asked whether DcR3 down-regulated host immunity by inducing immune cell apoptosis. We demonstrate that DcR3 induces dendritic cell (DC) apoptosis by activating PKC-delta and JNK subsequently to up-regulate DR5 to recruit Fas-associated death domain (FADD) to propagate the apoptotic signals. The association of FADD with DR5 results in the formation of death-inducing signaling complex (DISC) to trigger the downstream apoptotic signaling cascade. PKC-delta is activated via cross-linking of heparan sulfate proteoglycan (HSPG) on DCs, because recombinant protein containing the heparin-binding domain (HBD) of DcR3 and the Fc portion of IgG(1), the HBD.Fc fusion protein, is also able to trigger DC apoptosis. This provides the first evidence that cross-linking of HSPG on DCs can activate PKC-delta to induce DC apoptosis via the formation of DR5 DISC, and elucidates a novel mechanism of DcR3-mediated immunosuppression.


Assuntos
Apoptose , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Membro 6b de Receptores do Fator de Necrose Tumoral/metabolismo , Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Ativação Enzimática , Heparina/imunologia , Heparina/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ligação Proteica , Proteína Quinase C/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Membro 6b de Receptores do Fator de Necrose Tumoral/imunologia , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Regulação para Cima , Receptor fas/metabolismo
14.
J Immunol ; 176(1): 173-80, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16365408

RESUMO

Decoy receptor 3 (DcR3), a soluble receptor for Fas ligand, LIGHT (homologous to lymphotoxins shows inducible expression and competes with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes), and TNF-like molecule 1A, is highly expressed in cancer cells and in tissues affected by autoimmune disease. DcR3.Fc has been shown to stimulate cell adhesion and to modulate cell activation and differentiation by triggering multiple signaling cascades that are independent of its three known ligands. In this study we found that DcR3.Fc-induced cell adhesion was inhibited by heparin and heparan sulfate, and that DcR3.Fc was unable to bind Chinese hamster ovary K1 mutants defective in glycosaminoglycan (GAG) synthesis. Furthermore, the negatively charged, sulfated GAGs of cell surface proteoglycans, but not their core proteins, were identified as the binding sites for DcR3.Fc. A potential GAG-binding site was found in the C-terminal region of DcR3, and the mutation of three basic residues, i.e., K256, R258, and R259, to alanines abolished its ability to trigger cell adhesion. Moreover, a fusion protein comprising the GAG-binding region of DcR3 with an Fc fragment (DcR3_HBD.Fc) has the same effect as DcR3.Fc in activating protein kinase C and inducing cell adhesion. Compared with wild-type THP-1 cells, cell adhesion induced by DcR3.Fc was significantly reduced in both CD44v3 and syndecan-2 knockdown THP-1 cells. Therefore, we propose a model in which DcR3.Fc may bind to and cross-link proteoglycans to induce monocyte adhesion.


Assuntos
Glicosaminoglicanos/química , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Monócitos/imunologia , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/imunologia , Animais , Sequência de Bases , Sítios de Ligação , Células CHO , Adesão Celular , Cricetinae , Citometria de Fluxo , Quinase 1 de Adesão Focal/metabolismo , Glicosaminoglicanos/deficiência , Humanos , Dados de Sequência Molecular , Mutação , Reação em Cadeia da Polimerase , Proteína Quinase C/metabolismo , Membro 6b de Receptores do Fator de Necrose Tumoral , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
15.
J Immunol ; 175(8): 5135-45, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16210617

RESUMO

The soluble decoy receptor 3 (DcR3) is a member of the TNFR superfamily. Because DcR3 is up-regulated in tumor tissues and is detectable in the sera of cancer patients, it is regarded as an immunosuppressor to down-regulate immune responses. To understand the function of DcR3 in vivo, we generated transgenic mice overexpressing DcR3 systemically. In comparison with HNT-TCR (HNT) transgenic mice, up-regulation of IL-4 and IL-10 and down-regulation of IFN-gamma, IL-12, and TNF-alpha were observed in the influenza hemagglutinin(126-138) peptide-stimulated splenocytes of HNT-DcR3 double-transgenic mice. When infected with Listeria monocytogenes, DcR3 transgenic mice show attenuated expression of IFN-gamma as well as increased susceptibility to infection. The Th2 cell-biased phenotype in DcR3 transgenic mice is attributed to decreased IL-2 secretion by T cells, resulting in the suppression of IL-2 dependent CD4(+) T cell proliferation. This suggests that DcR3 might help tumor growth by attenuating the Th1 response and suppressing cell-mediated immunity.


Assuntos
Adjuvantes Imunológicos/genética , Ativação Linfocitária/genética , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Adjuvantes Imunológicos/metabolismo , Adjuvantes Imunológicos/fisiologia , Animais , Apoptose/fisiologia , Células Cultivadas , Citocinas/metabolismo , Proteína Ligante Fas , Humanos , Imunidade Celular/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos MRL lpr , Camundongos Transgênicos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/fisiologia , Membro 6b de Receptores do Fator de Necrose Tumoral , Células Th2/imunologia , Células Th2/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Fatores de Necrose Tumoral/metabolismo
16.
J Biol Chem ; 279(42): 44211-8, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15475369

RESUMO

Decoy receptor 3 (DcR3)/TR6/M68 is a soluble receptor that binds to the Fas ligand LIGHT and TL1A. Elevated levels of DcR3 expression have been found in many tumors. We report an unexpected effect of DcR3 by sensitizing Jurkat and U937 cells to apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Cell death triggered by anti-Fas and tumor necrosis factor was unaffected by DcR3. DcR3 by itself did not stimulate apoptosis. The ability to augment TRAIL-initiated cell death was not observed with soluble lymphotoxin beta receptor or soluble death receptor 3, indicating that binding to LIGHT or TL1A alone is insufficient to trigger TRAIL sensitivity. Incubation with DcR3 did not increase the surface expression of TRAIL receptor, and the level of Fas-associated death domain protein and cellular FLICE-like inhibitory protein was not altered. Instead, in the presence of DcR3, TRAIL engagement resulted in an increased activation of caspase-8, an elevated cleavage of Bid, and enhanced release of Smac and cytochrome c from mitochondria to cytosol compared with TRAIL alone. This led to increased activation of caspase-9 and caspase-3. The unusual ability of DcR3 to promote TRAIL-triggered death may be used to potentiate TRAIL efficacy during treatment tumors overexpressing DcR3.


Assuntos
Apoptose/fisiologia , Glicoproteínas de Membrana/fisiologia , Receptores de Superfície Celular/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Reguladoras de Apoptose , Morte Celular , Humanos , Células Jurkat , Receptores do Fator de Necrose Tumoral , Membro 6b de Receptores do Fator de Necrose Tumoral , Ligante Indutor de Apoptose Relacionado a TNF , Células U937 , Receptor fas/análise
17.
J Leukoc Biol ; 75(3): 486-94, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14657214

RESUMO

Decoy receptor 3 (DcR3) is a soluble receptor of the tumor necrosis factor receptor superfamily and is readily detected in certain cancer patients. Recently, we demonstrated that DcR3.Fc-treated dendritic cells skew T cell responses to a T helper cell type 2 phenotype. In this study, we further asked its ability to modulate CD14+ monocyte differentiation into macrophages induced by macrophage-colony stimulating factor in vitro. We found that DcR3.Fc was able to modulate the expression of several macrophage markers, including CD14, CD16, CD64, and human leukocyte antigen-DR. In contrast, the expression of CD11c, CD36, CD68, and CD206 (mannose receptor) was not affected in the in vitro culture system. Moreover, phagocytic activity toward immune complexes and apoptotic bodies as well as the production of free radicals and proinflammatory cytokines in response to lipopolysaccharide were impaired in DcR3.Fc-treated monocyte-derived macrophages. This suggests that DcR3.Fc might have potent, suppressive effects to down-regulate the host-immune system.


Assuntos
Macrófagos/efeitos dos fármacos , Glicoproteínas de Membrana/farmacologia , Antígenos CD/análise , Diferenciação Celular/efeitos dos fármacos , Interações Medicamentosas , Radicais Livres/metabolismo , Humanos , Células Jurkat , Receptores de Lipopolissacarídeos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/citologia , Macrófagos/imunologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Receptores de Superfície Celular , Receptores do Fator de Necrose Tumoral , Membro 6b de Receptores do Fator de Necrose Tumoral , Proteínas Recombinantes de Fusão/farmacologia
18.
Exp Cell Res ; 292(2): 241-51, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14697332

RESUMO

Decoy receptor 3 (DcR3), a newly identified soluble protein belonging to the tumor necrosis factor receptor (TNFR) superfamily, is a receptor for Fas ligand (FasL), LIGHT and TL1A. It has been demonstrated that DcR3 is frequently overexpressed by malignant tumors arising from lung, gastrointestinal tract, neuronal glia and virus-associated leukemia. Recently, we demonstrated that DcR3 is able to modulate the differentiation and activation of dendritic cells (DCs), and that DcR3-treated DCs skew naive T cell differentiation towards a Th2 phenotype. In this study, we further demonstrate that DcR3 is able to induce actin reorganization and enhance the adhesion of monocytes and THP-1 cells by activating multiple signaling molecules, such as protein kinase C (PKC), phosphatidylinositol 3-kinase (PI3K), focal adhesion kinase (FAK) and Src kinases. This provides the first evidence that the soluble DcR3, like other immobilized members of TNFR superfamily, is able to trigger 'reverse signaling' to modulate cell function.


Assuntos
Moléculas de Adesão Celular/metabolismo , Glicoproteínas de Membrana/metabolismo , Monócitos/metabolismo , Proteínas Serina-Treonina Quinases , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Actinas/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Glicoproteínas de Membrana/farmacologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptores do Fator de Necrose Tumoral , Membro 6b de Receptores do Fator de Necrose Tumoral , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
19.
J Immunol ; 168(10): 4846-53, 2002 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11994433

RESUMO

Decoy receptor 3 (DcR3), a soluble receptor belonging to the TNFR superfamily, is a receptor for both Fas ligand (FasL) and LIGHT. It has been demonstrated that DcR3 is up-regulated in lung and colon cancers, thus promoting tumor growth by neutralizing the cytotoxic effects of FasL and LIGHT. In this study, we found that DcR3.Fc profoundly modulated dendritic cell differentiation and maturation from CD14(+) monocytes, including the up-regulation of CD86/B7.2, and the down-regulation of CD40, CD54/ICAM-1, CD80/B7.1, CD1a, and HLA-DR. Moreover, DcR3-treated dendritic cells suppressed CD4(+) T cell proliferation in an allogeneic MLR and up-regulated IL-4 secretion of CD4(+)CD45RA(+) T cells. This suggests that DcR3.Fc may act not only as a decoy receptor to FasL and LIGHT, but also as an effector molecule to skew T cell response to the Th2 phenotype.


Assuntos
Adjuvantes Imunológicos/fisiologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Receptores de Superfície Celular/fisiologia , Adjuvantes Imunológicos/metabolismo , Animais , Células COS , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/metabolismo , Proteína Ligante Fas , Células HT29 , Humanos , Células Jurkat , Ligantes , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores do Fator de Necrose Tumoral/fisiologia , Membro 6b de Receptores do Fator de Necrose Tumoral , Células Th2/imunologia , Células Th2/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/metabolismo , Receptor fas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA